Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 127
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Immunity ; 56(3): 592-605.e8, 2023 03 14.
Article in English | MEDLINE | ID: mdl-36804959

ABSTRACT

Plasmodium replicates within the liver prior to reaching the bloodstream and infecting red blood cells. Because clinical manifestations of malaria only arise during the blood stage of infection, a perception exists that liver infection does not impact disease pathology. By developing a murine model where the liver and blood stages of infection are uncoupled, we showed that the integration of signals from both stages dictated mortality outcomes. This dichotomy relied on liver stage-dependent activation of Vγ4+ γδ T cells. Subsequent blood stage parasite loads dictated their cytokine profiles, where low parasite loads preferentially expanded IL-17-producing γδ T cells. IL-17 drove extra-medullary erythropoiesis and concomitant reticulocytosis, which protected mice from lethal experimental cerebral malaria (ECM). Adoptive transfer of erythroid precursors could rescue mice from ECM. Modeling of γδ T cell dynamics suggests that this protective mechanism may be key for the establishment of naturally acquired malaria immunity among frequently exposed individuals.


Subject(s)
Erythropoiesis , Malaria, Cerebral , Animals , Mice , Erythrocytes , Interleukin-17 , Liver/parasitology , Mice, Inbred C57BL , Receptors, Antigen, T-Cell, gamma-delta , Malaria
2.
Nature ; 611(7936): 563-569, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36352220

ABSTRACT

Malaria infection involves an obligatory, yet clinically silent liver stage1,2. Hepatocytes operate in repeating units termed lobules, exhibiting heterogeneous gene expression patterns along the lobule axis3, but the effects of hepatocyte zonation on parasite development at the molecular level remain unknown. Here we combine single-cell RNA sequencing4 and single-molecule transcript imaging5 to characterize the host and parasite temporal expression programmes in a zonally controlled manner for the rodent malaria parasite Plasmodium berghei ANKA. We identify differences in parasite gene expression in distinct zones, including potentially co-adaptive programmes related to iron and fatty acid metabolism. We find that parasites develop more rapidly in the pericentral lobule zones and identify a subpopulation of periportally biased hepatocytes that harbour abortive infections, reduced levels of Plasmodium transcripts and parasitophorous vacuole breakdown. These 'abortive hepatocytes', which appear predominantly with high parasite inoculum, upregulate immune recruitment and key signalling programmes. Our study provides a resource for understanding the liver stage of Plasmodium infection at high spatial resolution and highlights the heterogeneous behaviour of both the parasite and the host hepatocyte.


Subject(s)
Gene Expression Regulation , Hepatocytes , Liver , Malaria , Parasites , Plasmodium berghei , Single-Cell Analysis , Animals , Hepatocytes/cytology , Hepatocytes/immunology , Hepatocytes/metabolism , Hepatocytes/parasitology , Liver/anatomy & histology , Liver/cytology , Liver/immunology , Liver/parasitology , Malaria/genetics , Malaria/immunology , Malaria/parasitology , Parasites/genetics , Parasites/immunology , Parasites/metabolism , Plasmodium berghei/genetics , Plasmodium berghei/immunology , Plasmodium berghei/metabolism , Single Molecule Imaging , Sequence Analysis, RNA , Iron/metabolism , Fatty Acids/metabolism , Transcription, Genetic , Genes, Protozoan/genetics , Host-Parasite Interactions/genetics , Host-Parasite Interactions/immunology
3.
Eur J Immunol ; 52(1): 149-160, 2022 01.
Article in English | MEDLINE | ID: mdl-34695227

ABSTRACT

During the COVID-19 pandemic, Portugal has experienced three distinct SARS-CoV-2 infection waves. We previously documented the prevalence of SARS-CoV-2 immunity, measured by specific antibodies, in September 2020, 6 months after the initial moderate wave. Here, we show the seroprevalence changes 6 months later, up to the second week of March 2021, shortly following the third wave, which was one of the most severe in the world, and 2 months following the start of the vaccination campaign. A longitudinal epidemiological study was conducted, with a stratified quota sample of the Portuguese population. Serological testing was performed, including ELISA determination of antibody class and titers. The proportion of seropositives, which was 2.2% in September 2020, rose sharply to 17.3% (95% CI: 15.8-18.8%) in March 2021. Importantly, circulating IgG and IgA antibody levels were very stable 6 months after the initial determination and up to a year after initial infection, indicating long-lasting infection immunity against SARS-CoV-2. Moreover, vaccinated people had higher IgG levels from 3 weeks post-vaccination when compared with previously infected people at the same time post-infection.


Subject(s)
Antibodies, Viral/immunology , COVID-19 Serological Testing , COVID-19 , Immunoglobulin A/immunology , Immunoglobulin G/immunology , SARS-CoV-2/immunology , Adolescent , Adult , COVID-19/epidemiology , COVID-19/immunology , Female , Follow-Up Studies , Humans , Male , Middle Aged , Portugal/epidemiology , Prevalence , Time Factors
4.
Nature ; 547(7662): 213-216, 2017 07 13.
Article in English | MEDLINE | ID: mdl-28678779

ABSTRACT

The lifestyle of intracellular pathogens, such as malaria parasites, is intimately connected to that of their host, primarily for nutrient supply. Nutrients act not only as primary sources of energy but also as regulators of gene expression, metabolism and growth, through various signalling networks that enable cells to sense and adapt to varying environmental conditions. Canonical nutrient-sensing pathways are presumed to be absent from the causative agent of malaria, Plasmodium, thus raising the question of whether these parasites can sense and cope with fluctuations in host nutrient levels. Here we show that Plasmodium blood-stage parasites actively respond to host dietary calorie alterations through rearrangement of their transcriptome accompanied by substantial adjustment of their multiplication rate. A kinome analysis combined with chemical and genetic approaches identified KIN as a critical regulator that mediates sensing of nutrients and controls a transcriptional response to the host nutritional status. KIN shares homology with SNF1/AMPKα, and yeast complementation studies suggest that it is part of a functionally conserved cellular energy-sensing pathway. Overall, these findings reveal a key parasite nutrient-sensing mechanism that is critical for modulating parasite replication and virulence.


Subject(s)
Gene Expression Regulation , Malaria/parasitology , Parasites/metabolism , Parasites/pathogenicity , Phosphotransferases/metabolism , Plasmodium/metabolism , Plasmodium/pathogenicity , Animals , Caloric Restriction , Energy Metabolism/drug effects , Energy Metabolism/genetics , Gene Expression Regulation/drug effects , Genetic Complementation Test , Glucose/metabolism , Glucose/pharmacology , Male , Mice , Mice, Inbred C57BL , Parasitemia/blood , Parasitemia/genetics , Parasitemia/metabolism , Parasitemia/parasitology , Parasites/genetics , Parasites/growth & development , Phosphotransferases/deficiency , Phosphotransferases/genetics , Plasmodium/genetics , Plasmodium/growth & development , Rats , Transcriptome/drug effects , Virulence/drug effects
5.
Proc Natl Acad Sci U S A ; 116(20): 9979-9988, 2019 05 14.
Article in English | MEDLINE | ID: mdl-31028144

ABSTRACT

Cerebral malaria (CM) is a major cause of death due to Plasmodium infection. Both parasite and host factors contribute to the onset of CM, but the precise cellular and molecular mechanisms that contribute to its pathogenesis remain poorly characterized. Unlike conventional αß-T cells, previous studies on murine γδ-T cells failed to identify a nonredundant role for this T cell subset in experimental cerebral malaria (ECM). Here we show that mice lacking γδ-T cells are resistant to ECM when infected with Plasmodium berghei ANKA sporozoites, the liver-infective form of the parasite and the natural route of infection, in contrast with their susceptible phenotype if challenged with P. berghei ANKA-infected red blood cells that bypass the liver stage of infection. Strikingly, the presence of γδ-T cells enhanced the expression of Plasmodium immunogenic factors and exacerbated subsequent systemic and brain-infiltrating inflammatory αß-T cell responses. These phenomena were dependent on the proinflammatory cytokine IFN-γ, which was required during liver stage for modulation of the parasite transcriptome, as well as for downstream immune-mediated pathology. Our work reveals an unanticipated critical role of γδ-T cells in the development of ECM upon Plasmodium liver-stage infection.


Subject(s)
Intraepithelial Lymphocytes/physiology , Liver/immunology , Malaria, Cerebral/immunology , Plasmodium berghei/pathogenicity , Sporozoites/pathogenicity , Animals , Liver/parasitology , Male , Mice , Mice, Inbred C57BL , Sporozoites/growth & development
6.
Emerg Infect Dis ; 27(11): 2878-2881, 2021 11.
Article in English | MEDLINE | ID: mdl-34437830

ABSTRACT

In September 2020, we tested 13,398 persons in Portugal for antibodies against severe acute respiratory syndrome coronavirus 2 by using a quota sample stratified by age and population density. We found a seroprevalence of 2.2%, 3-4 times larger than the official number of cases at the end of the first wave of the pandemic.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , Pandemics , Portugal/epidemiology , Prevalence , Seroepidemiologic Studies
7.
Proc Natl Acad Sci U S A ; 114(7): E1138-E1147, 2017 02 14.
Article in English | MEDLINE | ID: mdl-28137845

ABSTRACT

The first, obligatory replication phase of malaria parasite infections is characterized by rapid expansion and differentiation of single parasites in liver cells, resulting in the formation and release of thousands of invasive merozoites into the bloodstream. Hepatic Plasmodium development occurs inside a specialized membranous compartment termed the parasitophorous vacuole (PV). Here, we show that, during the parasite's hepatic replication, the C-terminal region of the parasitic PV membrane protein exported protein 1 (EXP-1) binds to host Apolipoprotein H (ApoH) and that this molecular interaction plays a pivotal role for successful Plasmodium liver-stage development. Expression of a truncated EXP-1 protein, missing the specific ApoH interaction site, or down-regulation of ApoH expression in either hepatic cells or mouse livers by RNA interference resulted in impaired intrahepatic development. Furthermore, infection of mice with sporozoites expressing a truncated version of EXP-1 resulted in both a significant reduction of liver burden and delayed blood-stage patency, leading to a disease outcome different from that generally induced by infection with wild-type parasites. This study identifies a host-parasite protein interaction during the hepatic stage of infection by Plasmodium parasites. The identification of such vital interactions may hold potential toward the development of novel malaria prevention strategies.


Subject(s)
Liver/parasitology , Malaria/parasitology , Membrane Proteins/metabolism , Plasmodium berghei/physiology , Protozoan Proteins/metabolism , beta 2-Glycoprotein I/metabolism , Animals , Animals, Genetically Modified , Binding Sites , Down-Regulation , Genes, Protozoan , HEK293 Cells , Hepatocytes/parasitology , Humans , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Plasmodium berghei/genetics , Plasmodium berghei/growth & development , Protein Binding , RNA Interference , RNA, Small Interfering/genetics , Sequence Deletion , Sporozoites/physiology , Vacuoles/parasitology , beta 2-Glycoprotein I/antagonists & inhibitors , beta 2-Glycoprotein I/genetics
8.
Mol Microbiol ; 109(4): 458-473, 2018 08.
Article in English | MEDLINE | ID: mdl-29873127

ABSTRACT

Transmission of the malaria parasite Plasmodium falciparum involves infection of Anopheles mosquitoes. Here we characterize SOPT, a protein expressed in P. falciparum ookinetes that facilitates infection of the mosquito midgut. SOPT was identified on the basis that it contains a signal peptide, a PEXEL-like sequence and is expressed in asexual, ookinete and sporozoite stages, suggesting it is involved in infecting the human or mosquito host. SOPT is predicted to contain a subtilisin-like fold with a non-canonical catalytic triad and is orthologous to P. berghei PIMMS2. Localization studies reveal that SOPT is not exported to the erythrocyte but is expressed in ookinetes at the parasite periphery. SOPT-deficient parasites develop normally through the asexual and sexual stages and produce equivalent numbers of ookinetes to NF54 controls, however, they form fewer oocysts and sporozoites in mosquitoes. SOPT-deficient parasites were also unable to activate the immune-responsive midgut invasion marker SRPN6 after mosquito ingestion, suggesting they are defective for entry into the midgut. Disruption of SOPT in P. berghei (PIMMS2) did not affect other lifecycle stages or ookinete development but again resulted in fewer oocysts and sporozoites in mosquitoes. Collectively, this study shows that SOPT/PIMMS2 plays a conserved role in ookinetes of different Plasmodium species.


Subject(s)
Anopheles/parasitology , Digestive System/parasitology , Oocysts/growth & development , Plasmodium falciparum/pathogenicity , Protozoan Proteins/metabolism , Sporozoites/growth & development , Animals , Malaria, Falciparum/transmission , Mosquito Vectors/parasitology , Subtilisin/metabolism
9.
Cell Microbiol ; 18(3): 437-50, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26399761

ABSTRACT

Autophagy plays an important role in the defence against intracellular pathogens. However, some microorganisms can manipulate this host cell pathway to their advantage. In this study, we addressed the role of host cell autophagy during Plasmodium berghei liver infection. We show that vesicles containing the autophagic marker LC3 surround parasites from early time-points after invasion and throughout infection and colocalize with the parasitophorous vacuole membrane. Moreover, we show that the LC3-positive vesicles that surround Plasmodium parasites are amphisomes that converge from the endocytic and autophagic pathways, because they contain markers of both pathways. When the host autophagic pathway was inhibited by silencing several of its key regulators such as LC3, Beclin1, Vps34 or Atg5, we observed a reduction in parasite size. We also found that LC3 surrounds parasites in vivo and that parasite load is diminished in a mouse model deficient for autophagy. Together, these results show the importance of the host autophagic pathway for parasite development during the liver stage of Plasmodium infection.


Subject(s)
Autophagy/physiology , Host-Parasite Interactions/physiology , Liver/parasitology , Malaria/pathology , Plasmodium berghei/pathogenicity , Animals , Apoptosis Regulatory Proteins/metabolism , Beclin-1 , Liver/pathology , Malaria/parasitology , Mice, Inbred C57BL , Mice, Transgenic , Microtubule-Associated Proteins/metabolism
10.
EMBO Rep ; 16(8): 955-64, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26113366

ABSTRACT

Upon infection of a mammalian host, Plasmodium parasites first replicate inside hepatocytes, generating thousands of new parasites. Although Plasmodium intra-hepatic development represents a substantial metabolic challenge to the host hepatocyte, how infected cells respond to and integrate this stress remains poorly understood. Here, we present proteomic and transcriptomic analyses, revealing that the endoplasmic reticulum (ER)-resident unfolded protein response (UPR) is activated in host hepatocytes upon Plasmodium berghei infection. The expression of XBP1s--the active form of the UPR mediator XBP1--and the liver-specific UPR mediator CREBH is induced by P. berghei infection in vivo. Furthermore, this UPR induction increases parasite liver burden. Altogether, our data suggest that ER stress is a central feature of P. berghei intra-hepatic development, contributing to the success of infection.


Subject(s)
Endoplasmic Reticulum Stress , Hepatocytes/parasitology , Host-Parasite Interactions , Malaria/parasitology , Plasmodium berghei/growth & development , Unfolded Protein Response , Animals , DNA-Binding Proteins/genetics , Gene Expression Profiling , Hepatocytes/physiology , Hepatocytes/ultrastructure , Life Cycle Stages , Malaria/physiopathology , Male , Mice, Inbred C57BL , Parasite Load , Plasmodium berghei/pathogenicity , Proteomics , Regulatory Factor X Transcription Factors , Signal Transduction/genetics , Transcription Factors/genetics , X-Box Binding Protein 1
11.
Eukaryot Cell ; 14(1): 96-103, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25416236

ABSTRACT

Prior to invading nonreplicative erythrocytes, Plasmodium parasites undergo their first obligate step in the mammalian host inside hepatocytes, where each sporozoite replicates to generate thousands of merozoites. While normally quiescent, hepatocytes retain proliferative capacity and can readily reenter the cell cycle in response to diverse stimuli. Many intracellular pathogens, including protozoan parasites, manipulate the cell cycle progression of their host cells for their own benefit, but it is not known whether the hepatocyte cell cycle plays a role during Plasmodium liver stage infection. Here, we show that Plasmodium parasites can be observed in mitotic hepatoma cells throughout liver stage development, where they initially reduce the likelihood of mitosis and ultimately lead to significant acquisition of a binucleate phenotype. However, hepatoma cells pharmacologically arrested in S phase still support robust and complete Plasmodium liver stage development, which thus does not require cell cycle progression in the infected cell in vitro. Furthermore, murine hepatocytes remain quiescent throughout in vivo infection with either Plasmodium berghei or Plasmodium yoelii, as do Plasmodium falciparum-infected primary human hepatocytes, demonstrating that the rapid and prodigious growth of liver stage parasites is accomplished independent of host hepatocyte cell cycle progression during natural infection.


Subject(s)
Hepatocytes/physiology , Mitosis , Plasmodium/growth & development , Sporozoites/physiology , Animals , Cell Nucleus/physiology , Hep G2 Cells , Hepatocytes/cytology , Hepatocytes/parasitology , Host-Parasite Interactions , Humans , Mice , Mice, Inbred C57BL , Plasmodium/pathogenicity
12.
Proc Natl Acad Sci U S A ; 110(30): E2838-47, 2013 Jul 23.
Article in English | MEDLINE | ID: mdl-23836641

ABSTRACT

Residence within a customized vacuole is a highly successful strategy used by diverse intracellular microorganisms. The parasitophorous vacuole membrane (PVM) is the critical interface between Plasmodium parasites and their possibly hostile, yet ultimately sustaining, host cell environment. We show that torins, developed as ATP-competitive mammalian target of rapamycin (mTOR) kinase inhibitors, are fast-acting antiplasmodial compounds that unexpectedly target the parasite directly, blocking the dynamic trafficking of the Plasmodium proteins exported protein 1 (EXP1) and upregulated in sporozoites 4 (UIS4) to the liver stage PVM and leading to efficient parasite elimination by the hepatocyte. Torin2 has single-digit, or lower, nanomolar potency in both liver and blood stages of infection in vitro and is likewise effective against both stages in vivo, with a single oral dose sufficient to clear liver stage infection. Parasite elimination and perturbed trafficking of liver stage PVM-resident proteins are both specific aspects of torin-mediated Plasmodium liver stage inhibition, indicating that torins have a distinct mode of action compared with currently used antimalarials.


Subject(s)
Antimalarials/pharmacology , Liver/parasitology , Membrane Proteins/metabolism , Naphthyridines/pharmacology , Plasmodium/drug effects , Animals , Plasmodium/metabolism , Vacuoles/metabolism
13.
Infect Immun ; 83(3): 1172-80, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25583524

ABSTRACT

Following transmission through a mosquito bite to the mammalian host, Plasmodium parasites first invade and replicate inside hepatocytes before infecting erythrocytes and causing malaria. The mechanisms limiting Plasmodium reinfections in humans living in regions of malaria endemicity have mainly been explored by studying the resistance induced by the blood stage of infection. However, epidemiologic studies have suggested that in high-transmission areas, preerythrocytic stages also activate host resistance to reinfection. This, along with the recent discovery that liver infections trigger a specific and effective type I interferon (IFN) response, prompted us to hypothesize that this pre-erythrocyte-stage-induced resistance is linked to liver innate immunity. Here, we combined experimental approaches and mathematical modeling to recapitulate field studies and understand the molecular basis behind such resistance. We present a newly established mouse reinfection model and demonstrate that rodent malaria liver-stage infection inhibits reinfection. This protection relies on the activation of innate immunity and involves the type I IFN response and the antimicrobial cytokine gamma IFN (IFN-γ). Importantly, mathematical simulations indicate that the predictions based on our experimental murine reinfection model fit available epidemiological data. Overall, our study revealed that liver-stage-induced innate immunity may contribute to the preerythrocytic resistance observed in humans in regions of malaria hyperendemicity.


Subject(s)
Adaptive Immunity , Liver/immunology , Malaria/immunology , Models, Statistical , Plasmodium berghei/immunology , Sporozoites/immunology , Adaptor Proteins, Signal Transducing , Animals , Anopheles/parasitology , Carrier Proteins/genetics , Carrier Proteins/immunology , Gene Expression Regulation/immunology , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Immunity, Innate , Immunologic Memory , Interferon Regulatory Factor-7/genetics , Interferon Regulatory Factor-7/immunology , Interferon-gamma/deficiency , Interferon-gamma/genetics , Interferon-gamma/immunology , Intracellular Signaling Peptides and Proteins , Liver/parasitology , Malaria/genetics , Malaria/parasitology , Mice , Mice, Inbred C57BL , Mice, Knockout , Parasite Load , Plasmodium berghei/growth & development , Proteins/genetics , Proteins/immunology , RNA-Binding Proteins , Receptor, Interferon alpha-beta/deficiency , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/immunology , Sporozoites/growth & development , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/immunology
14.
J Nanobiotechnology ; 13: 48, 2015 Aug 07.
Article in English | MEDLINE | ID: mdl-26250828

ABSTRACT

BACKGROUND: Gold nanoparticles have been widely employed for biosensing purposes with remarkable efficacy for DNA detection. Amongst the proposed systems, colorimetric strategies based on the remarkable optical properties have provided for simple yet effective sequence discrimination with potential for molecular diagnostics at point of need. These systems may also been used for parallel detection of several targets to provide additional information on diagnostics of pathogens. RESULTS: For the first time, we demonstrate that a single Au-nanoprobe may provide for detection of two distinct targets (pathogens) allowing colorimetric multi-target detection. We demonstrate this concept by using one single gold-nanoprobe capable to detect members of the Mycobacterium tuberculosis complex and Plasmodium sp., the etiologic agents of tuberculosis and malaria, respectively. Following characterisation, the developed gold-nanoprobe allowed detection of either target in individual samples or in samples containing both DNA species with the same efficacy. CONCLUSIONS: Using one single probe via the non-cross-linking colorimetric methodology it is possible to identify multiple targets in one sample in one reaction. This proof-of-concept approach may easily be integrated into sensing platforms allowing for fast and simple multiplexing of Au-nanoprobe based detection at point-of-need.


Subject(s)
Gold/chemistry , Malaria/diagnosis , Metal Nanoparticles/chemistry , Mycobacterium tuberculosis/isolation & purification , Plasmodium/isolation & purification , Tuberculosis/diagnosis , Colorimetry/methods , DNA, Bacterial/analysis , DNA, Bacterial/genetics , DNA, Protozoan/analysis , DNA, Protozoan/genetics , Humans , Malaria/parasitology , Nanotechnology/methods , Plasmodium/genetics , Point-of-Care Systems , Tuberculosis/microbiology
15.
Proc Natl Acad Sci U S A ; 109(22): 8511-6, 2012 May 29.
Article in English | MEDLINE | ID: mdl-22586124

ABSTRACT

Human malaria infection begins with a one-time asymptomatic liver stage followed by a cyclic symptomatic blood stage. All high-throughput malaria drug discovery efforts have focused on the cyclic blood stage, which has limited potential for the prophylaxis, transmission blocking, and eradication efforts that will be needed in the future. To address these unmet needs, a high-throughput phenotypic liver-stage Plasmodium parasite screen was developed to systematically identify molecules with liver-stage efficacy. The screen recapitulates liver-stage infection by isolating luciferase-expressing Plasmodium berghei parasites directly from the salivary glands of infected mosquitoes, adding them to confluent human liver cells in 384-well plates, and measuring luciferase activity after a suitable incubation period. Screening 5,375 known bioactive compounds identified 37 liver-stage malaria inhibitors with diverse modes of action, as shown by inhibition time course experiments. Further analysis of the hits in the Food and Drug Administration-approved drug subset revealed compounds that seem to act specifically on the liver stage of infection, suggesting that this phase of the parasite's life cycle presents a promising area for new drug discovery. Notably, many active compounds in this screen have molecular structures and putative targets distinctly different from those of known antimalarial agents.


Subject(s)
Antimalarials/pharmacology , Liver/drug effects , Malaria/prevention & control , Plasmodium berghei/drug effects , Animals , Anopheles/parasitology , Antimalarials/classification , Drug Evaluation, Preclinical , Hep G2 Cells , Humans , Inhibitory Concentration 50 , Insect Vectors/parasitology , Life Cycle Stages , Liver/parasitology , Liver/pathology , Malaria/parasitology , Malaria, Falciparum/parasitology , Malaria, Falciparum/prevention & control , Male , Mice , Mice, Inbred C57BL , Plasmodium berghei/growth & development , Plasmodium berghei/isolation & purification , Plasmodium falciparum/drug effects , Plasmodium falciparum/growth & development , Treatment Outcome
16.
Chembiochem ; 15(13): 1920-30, 2014 Sep 05.
Article in English | MEDLINE | ID: mdl-25111632

ABSTRACT

Malaria, an infectious disease caused by eukaryotic parasites of the genus Plasmodium, afflicts hundreds of millions of people every year. Both the parasite and its host utilize protein kinases to regulate essential cellular processes. Bioinformatic analyses of parasite genomes predict at least 65 protein kinases, but their biological functions and therapeutic potential are largely unknown. We profiled 1358 small-molecule kinase inhibitors to evaluate the role of both the human and the malaria kinomes in Plasmodium infection of liver cells, the parasites' obligatory but transient developmental stage that precedes the symptomatic blood stage. The screen identified several small molecules that inhibit parasite load in liver cells, some with nanomolar efficacy, and each compound was subsequently assessed for activity against blood-stage malaria. Most of the screening hits inhibited both liver- and blood-stage malaria parasites, which have dissimilar gene expression profiles and infect different host cells. Evaluation of existing kinase activity profiling data for the library members suggests that several kinases are essential to malaria parasites, including cyclin-dependent kinases (CDKs), glycogen synthase kinases, and phosphoinositide-3-kinases. CDK inhibitors were found to bind to Plasmodium protein kinase 5, but it is likely that these compounds target multiple parasite kinases. The dual-stage inhibition of the identified kinase inhibitors makes them useful chemical probes and promising starting points for antimalarial development.


Subject(s)
Genome, Protozoan/genetics , Malaria/genetics , Plasmodium/genetics , Protein Kinases/genetics , Animals , Antimalarials/chemistry , Computational Biology , Drug Evaluation, Preclinical , Humans , Liver/parasitology , Malaria/parasitology , Male , Mice , Mice, Inbred C57BL , Plasmodium/enzymology , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacology , Small Molecule Libraries
17.
Malar J ; 13: 15, 2014 Jan 08.
Article in English | MEDLINE | ID: mdl-24400642

ABSTRACT

BACKGROUND: The first phase of malaria infection occurs in the liver and is clinically silent. Inside hepatocytes each Plasmodium sporozoite replicate into thousands of erythrocyte-infectious merozoites that when released into the blood stream result in clinical symptoms of the disease. The time between sporozoite inoculation and the appearance of parasites in the blood is defined as the pre-patent period, which is classically analysed by time-consuming and labor-intensive techniques, such as microscopy and PCR. METHODS: Luciferase-expressing Plasmodium berghei parasites were used to measure pre-patent period of malaria infection in rodents using a bioluminescence assay that requires only one microliter of blood collected from the tail-vein. The accuracy and sensitivity of this new method was compared with conventional microscopy and PCR based techniques, and its capacity to measure the impact of anti-malarial interventions against the liver evaluated. RESULTS: The described method is very sensitive allowing the detection of parasites during the first cycles of blood stage replication. It accurately translates differences in liver load due to inoculation of different sporozoite doses as well as a result of treatment with different primaquine regimens. CONCLUSIONS: A novel, simple, fast, and sensitive method to measure pre-patent period of malaria infection in rodents is described here. The sensitivity and accuracy of this new method is comparable to standard PCR and microscopy-based techniques, respectively.


Subject(s)
Luminescent Measurements/methods , Malaria/diagnosis , Plasmodium berghei/isolation & purification , Animals , Blood/parasitology , Luciferases/metabolism , Malaria/parasitology , Male , Mice , Mice, Inbred C57BL , Plasmodium berghei/genetics , Polymerase Chain Reaction , Statistics, Nonparametric
18.
Bioorg Med Chem Lett ; 24(17): 4151-7, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-25103602

ABSTRACT

The malaria parasite Plasmodium goes through two life stages in the human host, a non-symptomatic liver stage (LS) followed by a blood stage with all clinical manifestation of the disease. In this study, we investigated a series of 2-alkynoic fatty acids (2-AFAs) with chain lengths between 14 and 18 carbon atoms for dual in vitro activity against both life stages. 2-Octadecynoic acid (2-ODA) was identified as the best inhibitor of Plasmodium berghei parasites with ten times higher potency (IC50=0.34 µg/ml) than the control drug. In target determination studies, the same compound inhibited three Plasmodium falciparum FAS-II (PfFAS-II) elongation enzymes PfFabI, PfFabZ, and PfFabG with the lowest IC50 values (0.28-0.80 µg/ml, respectively). Molecular modeling studies provided insights into the molecular aspects underlying the inhibitory activity of this series of 2-AFAs and a likely explanation for the considerably different inhibition potentials. Blood stages of P. falciparum followed a similar trend where 2-ODA emerged as the most active compound, with 20 times less potency. The general toxicity and hepatotoxicity of 2-AFAs were evaluated by in vitro and in vivo methods in mammalian cell lines and zebrafish models, respectively. This study identifies 2-ODA as the most promising antiparasitic 2-AFA, particularly towards P. berghei parasites.


Subject(s)
Antimalarials/pharmacology , Fatty Acid Synthase, Type II/antagonists & inhibitors , Fatty Acids, Unsaturated/pharmacology , Malaria/drug therapy , Malaria/parasitology , Plasmodium berghei/enzymology , Plasmodium falciparum/enzymology , Animals , Antimalarials/chemical synthesis , Antimalarials/chemistry , Cell Line, Tumor , Dose-Response Relationship, Drug , Fatty Acid Synthase, Type II/metabolism , Fatty Acids, Unsaturated/chemical synthesis , Fatty Acids, Unsaturated/chemistry , Humans , Models, Molecular , Plasmodium berghei/drug effects , Plasmodium falciparum/drug effects , Structure-Activity Relationship , Zebrafish
19.
Nat Med ; 13(6): 703-10, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17496899

ABSTRACT

Cerebral malaria claims more than 1 million lives per year. We report that heme oxygenase-1 (HO-1, encoded by Hmox1) prevents the development of experimental cerebral malaria (ECM). BALB/c mice infected with Plasmodium berghei ANKA upregulated HO-1 expression and activity and did not develop ECM. Deletion of Hmox1 and inhibition of HO activity increased ECM incidence to 83% and 78%, respectively. HO-1 upregulation was lower in infected C57BL/6 compared to BALB/c mice, and all infected C57BL/6 mice developed ECM (100% incidence). Pharmacological induction of HO-1 and exposure to the end-product of HO-1 activity, carbon monoxide (CO), reduced ECM incidence in C57BL/6 mice to 10% and 0%, respectively. Whereas neither HO-1 nor CO affected parasitemia, both prevented blood-brain barrier (BBB) disruption, brain microvasculature congestion and neuroinflammation, including CD8(+) T-cell brain sequestration. These effects were mediated by the binding of CO to hemoglobin, preventing hemoglobin oxidation and the generation of free heme, a molecule that triggers ECM pathogenesis.


Subject(s)
Carbon Monoxide/physiology , Heme Oxygenase-1/physiology , Heme/metabolism , Malaria, Cerebral/enzymology , Animals , Disease Models, Animal , Heme Oxygenase-1/deficiency , Heme Oxygenase-1/genetics , Malaria, Cerebral/drug therapy , Malaria, Cerebral/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, SCID , Plasmodium berghei
20.
bioRxiv ; 2024 May 14.
Article in English | MEDLINE | ID: mdl-38798622

ABSTRACT

Malaria transmission begins when infected female Anopheles mosquitos deposit Plasmodium parasites into the mammalian host's skin during a bloodmeal. The salivary gland-resident sporozoite parasites migrate to the bloodstream, subsequently invading and replicating within hepatocytes. As Anopheles mosquitos are more active at night, with a 24-hour rhythm, we investigated whether their salivary glands are under circadian control, anticipating bloodmeals and modulating sporozoite biology for host encounters. Here we show that approximately half of the mosquito salivary gland transcriptome, particularly genes essential for efficient bloodmeals such as anti-blood clotting factors, exhibits circadian rhythmic expression. Furthermore, we demonstrate that mosquitoes prefer to feed during nighttime, with the amount of blood ingested varying cyclically throughout the day. Notably, we show a substantial subset of the sporozoite transcriptome cycling throughout the day. These include genes involved in parasite motility, potentially modulating the ability to initiate infection at different times of day. Thus, although sporozoites are typically considered quiescent, our results demonstrate their transcriptional activity, revealing robust daily rhythms of gene expression. Our findings suggest a circadian evolutionary relationship between the vector, parasite and mammalian host that together modulate malaria transmission.

SELECTION OF CITATIONS
SEARCH DETAIL